EP0417191B1 - Formulierung für antikörper enthaltende reagenzien - Google Patents

Formulierung für antikörper enthaltende reagenzien Download PDF

Info

Publication number
EP0417191B1
EP0417191B1 EP89906915A EP89906915A EP0417191B1 EP 0417191 B1 EP0417191 B1 EP 0417191B1 EP 89906915 A EP89906915 A EP 89906915A EP 89906915 A EP89906915 A EP 89906915A EP 0417191 B1 EP0417191 B1 EP 0417191B1
Authority
EP
European Patent Office
Prior art keywords
maltose
buffer
antibody
sodium chloride
aqueous buffer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
EP89906915A
Other languages
English (en)
French (fr)
Other versions
EP0417191A1 (de
Inventor
David J. Shealey
Christopher P. Phillips
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Janssen Biotech Inc
Original Assignee
Centocor Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Centocor Inc filed Critical Centocor Inc
Priority to AT89906915T priority Critical patent/ATE86502T1/de
Publication of EP0417191A1 publication Critical patent/EP0417191A1/de
Application granted granted Critical
Publication of EP0417191B1 publication Critical patent/EP0417191B1/de
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39591Stabilisation, fragmentation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/02Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6887Antibody-chelate conjugates using chelates for therapeutic purposes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates generally to antibody preparations and particularly to a highly stabilized antibody preparation for parenteral administration.
  • HSA human serum albumin
  • U.S. Patent 4,186,192, to Lundblad et al. discloses the use of maltose to increase the stability of an immune serum globulin preparation for intramuscular or intravenous administration.
  • U.S. Patent 2,826,533 discloses the use of dextrose to increase the solubility of a fibrinogen preparation.
  • U.S. Patent 4,089,944 Thomas discloses the use of a variety of carbohydrates, such as dextrose, mannose, galactose, fructose, lactose, sucrose, and maltose to increase the solubility of an AHF-fibrinogen composition. It has been found that when dextrose is added to immune serum globulin to enhance stability and or solubility, the globulins tend to aggregate over time, thereby increasing the optical density of the solution, and resulting in shedding.
  • Shedding is an undesirable manifestation since it is visually observable and indicates the possibility that the shedded protein may be inactive or denatured, therefore reducing the effective amount of protein available.
  • a protein preparation in which shedding is apparent is an unsatisfactory product in terms of visual appearance.
  • maltose is readily available in pure form and has good stability in aqueous solutions in concentrations up to 20% by weight. Preparations containing maltose can be autoclaved without browning of the solution. Maltose in small quantities is practically physiologically inert. When administered parenterally, it is partially converted to glucose by the specific enzyme maltase found in many tissue sites in most animal species, including humans. The conversion to glucose is gradual and frequently undetectable when plasma glucose is serially measured; therefore there is no apparent increase in circulating insulin levels. Since maltose is a disaccharide, a 10% solution is approximately isotonic in humans. In U.S.
  • Patent 4,499,073 discloses the use of a carbohydrate in a preparation of immune serum globulin to impart physiologically acceptable isotonicity to the preparation. Tenold specifies 10% weight to volume maltose for this purpose. Lundblad et al., report in U.S. Patent 4,186,192 that a solution of immune serum globulin is stabilized when maltose is added in a concentration of between 5 and 18% by weight. Fernandes et al. describe a preparation of intravenous gamma-globulin stabilized with maltose to minimize precipitation and improve in vitro shelf stability. Vox Sang , 39 :101-112(1980).
  • the invention comprises an aqueous stabilizing buffer containing an antibody or antibody fragments, and maltose.
  • This buffer composition has the ability to inhibit the antibody or antibody fragments in solutions intended for intravenous administration from precipitating and forming particulates in the final product vial.
  • the antibody or antibody fragment may be derivatized with a chelating agent, such as, for example, diethylenetriamiepentaacetic acid (DTPA) for binding radiometals.
  • DTPA diethylenetriamiepentaacetic acid
  • the formulation of the buffer solution contains phosphate, sodium chloride and maltose.
  • the present composition has been successful in stabilizing monoclonal antibodies, or fragments thereof, for shipping and short-term storage at ambient temperatures without loss of immunoreactivity, and requires no refrigeration or other special handling.
  • the invention provides a stable, liquid formulation for monoclonal antibody products without shedding, thus increasing the shelf life of the antibody product.
  • the aqueous stabilizing buffer of this invention minimizes the formation of protein aggregates and particulates in reagents containing antibodies or antibody fragments, and insures that the antibody in solution maintains its immunoreactivity over time.
  • the preparation comprises a sterile, pharmaceutically acceptable solution containing a phosphate buffer, sodium chloride, an antimyosin monoclonal antibody or antibody fragment, and maltose.
  • a preferred embodiment of this invention comprises about 10mM to about 100mM sodium phosphate (pH 6-8), about 145mM sodium chloride and about 5-20% (w/v) maltose and between about 0.5-5.2 mg/ml antibody, preferably antimyosin.
  • other antibodies or fragments for example, antifibrin may be used.
  • This buffer enhances the stability of immunological activity of the monoclonal antibody, and prevents the immunoglobulins in solution intended for intravenous administration to human subjects from precipitating and forming particulates in the final product vial.
  • Another embodiment of the formulation contains pure monoclonal antibody molecules, or fragments, that have been modified for diagnostic therapeutic applications; for example derivatized with a chelating agent such as diethylenetriamine° pentaacetic acid (DTPA).
  • DTPA diethylenetriamine° pentaacetic acid
  • the derivatized antibody can then be used as a radiopharmaceutical due to the chelator's ability to bind a radioactive heavy metal, such as, for example, Indium-111.
  • the antibody solution includes a monoclonal antibody fragment, such as antimyosin, derivatized with DTPA.
  • the chelating agent is used for incorporating a radiometal, such as Indiun-111, into the antibody protein, forming a protein-chelate-radiometal complex. This complex is then administered to a subject to deliver the radiometal to a site defined by the antigen which is the target of the antibody.
  • the radiolabeled antibody can be used in scintigraphy, for example, in the imaging of tumors, or of disease sites, such as mycocardial infarct or blood clots.
  • injection of labeled antimyosin antibody which is specific for cardiac myosin, will result in localization of the radiometal at the site where antimyosin binds to myosin, and the site can then be scanned with a gamma camera to obtain an image of the myocardium useful for diagnostic purposes.
  • the present formulation exhibits superior stabilizing characteristics in terms of minimal protein particle formation, preservation of immunoreactivity and radiometal incorporation over time, and under stress conditions, such as elevated temperatures, vial filling and shipping.
  • Maltose which is used to stabilize the antibody solution, is described in detail in, for example, the Merck Index, 10th edition, Merck and Co., Inc. Rahway, NJ (1983).
  • Maltose is a disaccharide, (4-0- ⁇ -D-glucopyranosyl-D-glucopyranose), which has been established as useful for maintaining pharmaceutically acceptable isotonicity of immunoglobulin solutions. (See U.S. Patent 4,499,073 to Tenold and U.S. Patent 4,186,192 to Lundblad et al., both discussed hereinabove). It has also been determined that maltose is not metabolized by humans when administered intravenously, and is excreted as maltose, with no apparent elevation in blood glucose levels or release of insulin.
  • Buffers have long been used to solubilize and stabilize antibody products for parenteral injection. They are utilized as biologically acceptable carriers for proteins. Protein solubility in the buffer solution depends upon a number of factors, such as ionic strength of the solution and the isoelectric point of the protein. Buffers which have been used as antibody carriers include citrate, sodium chloride and phosphate. The preferred buffer for this formulation is sodium phosphate buffers.
  • Sodium chloride is added to antibody compositions to enhance stability and to render the solution physiologically acceptable upon injection.
  • Other alkali metal salts, such as potassium chloride, are not physiologically acceptable when injected intraveneously.
  • composition of the invention has successfully maintained the following characteristics after 65 weeks: antibody solubility (determined via liquid borne particulate analysis), chelator activity (greater than 88% binding of Indium-111 at 10 minutes), antibody immunoactivity (when compared to reference standard material) and antibody molecular integrity (via high pressure liquid chromatography and SDS page electrophoresis comparisons to reference standard material.
  • Formulations were tested with a variety of buffers, salt concentrations, pH levels, and excipients, such as, human serum albumin, surfactants, mandelic acid and N-acetyl tryptophanate.
  • the test formulations were initially screened by visual inspection after incubation at 4°C, 22°C, 37°C and 45°C.
  • test buffers Sodium citrate (Sigma Chemical Co., St. Louis, MO) Sodium chloride (J.T. Baker Co.) Sodium phosphate Monobasic and Dibasic (Sigma Chemical Co.) Maltose (Sigma Chemical Co.) Lactose (Sigma Chemical Co.) Tween® 80 (Sigma Chemical Co.) Dextrose (Sigma Chemical Co.) Human Serum Albumin (25%) (Armour) Propylene Glycol (Fisher Scientific Co.) Sodium acetate (Sigma Chemical Co.) Trishydroxymethylaminomethane (tris buffer) (Sigma Chemical Co.) Hydroxyethyl piperazine ethane sulfonic acid (HEPES buffer) (Sigma Chemical Co.) N-acetyl tryptophanate (Sigma Chemical Co.) Mandelic acid (Sigma Chemical Co.)
  • Antimyosin Fab-DTPA was dialyzed against 100 volumes of the indicated buffers using dialysis tubing (Fisher Scientific Co.), and then 0.2 ⁇ m filtered. Dialysis tubing was boiled in 10 mM EDTA pH 7.0, rinsed with distilled water, and stored in 70% ethanol prior to use.
  • Dialyzed antimyosin Fab-DTPA was adjusted to 0.5 mg/ml and filled, 1 ml/vial, aseptically into sterile 1 ml vials (Wheaton) and sealed with sterile rubber stoppers (West) and metal crimps. Vials were incubated at 4°C, 22°C, 37°C and 45°C for 48-96 hours. Some vials were, in addition, stressed by shaking at 37°C (formulations 1-32) or by shipping in styrofoam containers from Malvern, PA to Miami, FL, then returned to Malvern, PA by Federal Express, Priority 1 mail (formulations 90-95). Shipped vials were filled using a peristaltic pump (Paxall) through 0.123 inch (3.075 mm inside diameter) silicone tubing at a pump speed of 500 rpm.
  • Paxall peristaltic pump
  • Vials were visually inspected by inverting several times and observing against a dark and a light background. Vials containing antimyosin Fab-DTPA were compared with control vials, prepared as described above, which contained buffer only. In general, all of these buffers exhibited no shedding at 4°C and 22°C over the times they were examined. Experimental parameters for formulations exposed to temperatures of 37°C and 45°C yielded the most information. The degree of precipitation was graded as follows: + (precipitates, cloudy); +/- (fine precipitates); - (no precipitates). The results of the visual inspection for buffers 1-89 for formulations stressed at 37°C and 45°C are shown in Table 1.
  • In-111 labeled antimyosin Fab-DTPA After screening buffers for particulate formation, selected formulations which showed minimal precipitation were further evaluated for In-111 incorporation into the protein, and immunoreactivity of the resulting In-111 labeled antimyosin Fab-DTPA.
  • Formulations at neutral pH were acidified prior to radiolabeling with an equal volume of 0.2 M sodium citrate (pH 5) in a metal-free microfuge tube (BioRad). All transfers were also performed with metal-free pipette tips (BioRad).
  • In-111 chloride [Amersham, 370 MBq/ml (10 mCi/ml) at reference] was then added to the protein-citrate mixtures to a final specific activity of 148 MBq (4 mCi) per milligram.
  • a 1000-fold dilution of the radiolabeled antimyosin Fab-DTPA was made in 0.01 M sodium phosphate (pH 7.2), 0.15 M NaCl, 1% (w/v) bovine serum albumin (PBS-1% BSA).
  • PBS-1% BSA bovine serum albumin
  • One hundred microliters of this diluted sample were applied to a 1 ml column of myosin-Sepharose®C1-4B (Pharmacia). This affinity column was prepared by the attachment of myosin purified from dog heart tissue to cyanogen-bromide activated Sepharose® C1-4B (Pharmacia).
  • the column was eluted with eight 1 ml aliquots of PBS-1% BSA, followed by eight 1 ml aliquots of 0.1M glycine pH 2,5, 0.01% thimerosal.
  • the collected fractions were counted in a gamma counter set for In-111, the percentage eluting with the glycine buffer representing active radiolabeled antibody. This percentage was divided by the fraction of In-111 protein bound (from ITLC-SG chromatography) to correct for unbound In-111.
  • Table 4 shows an example with the excipient human serum albumin, where it appears that a portion of the In-111 binds to sites on the albumin rather than to the antimyosin antibody.
  • Neutral pH formulations require acidification prior to In-111 labeling. This was accomplished by adding an equal volume of a citrate buffer. Citrate buffers ranging from 100 mM to 500 mM and from pH 3.4 to 5.2 were equally effective. A 200 mM citrate pH 5.0 buffer was chosen for all future studies. Previous stability studies of antimyosin Fab-DTPA in 100 mM citrate pH 5.0 indicated no loss of immunoreactivity up to 66 weeks at 5°C.
  • formulation 90 which has the composition 10 mM phosphate, 145 mM NaCl, 10% maltose and containing 0.5 mg/ml antimyosin Fab-DTPA. This formulation, at pH 7.2, was pumped into vials and compared with the previous 100 mM citrate pH 5.0 formulation. The results are shown in Table 5:
  • the phosphate formulation (Formulation II) showed no particulates under these conditions, still bound In-111, and maintained a high level of immunoreactivity.
  • Vials of antimyosin Fab-DTPA (Centocor, Inc.) were stored under the following conditions: 4°C for 12 months ambient temperature for 12 months 37°C for 12 months and analyzed for particulates by particle counting and visual inspection.
  • All vials of antimyosin Fab-DTPA were filled at 1.15 ml/vial and contained 0.5 mg protein per ml of buffer solution.
  • the buffer solution was composed of 100 mM sodium phosphate (pH 6.5), 145 mM NaCl, 10% maltose.
  • the contents were then withdrawn by syringe through an 0.2 ⁇ m filter (Millipore® Millex-GV cat no. SLGVO25LS).
  • the syringe/filter/needle assembly and the filter/needle were weighed before and after filtration in order to measure the weight of the entire dose and the weight retained in the filter/needle.
  • the uCi of Indium-111 in the syringe/filter/needle and in the filter/needle was measured in a dose calibrator (Capintec CRC-5) after filtration.
  • the second stressed vial of antimyosin Fab-DTPA was opened in a laminar flow hood and analyzed on the particle counter. Each vial was opened and analyzed in a horizontal laminar flow hood. Three 0.1 ml aliquots were counted in a Climet model CI-1000 particle analyzer set to count all particles greater than or equal to 10 ⁇ m and all particles greater than or equal to 25 ⁇ m. USP XXI specifies that a single dose must contain 10,000 or fewer particles greater than or equal 10 ⁇ m in size and 1,000 or fewer particles greater than or equal to 25 ⁇ m in size. The remaining contents (about 0.5ml) were transferred to a centrifugal filter unit (Rainin cat.
  • SDS-PAGE Sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) using gradient gels and a discontinuous buffer system was carried out on the Pharmacia PhastSystem using PhastGel gradient 10-15 precast gels and PhastGel SDS buffer strips.
  • the 0.5 mm thick gels have a 4.5% T (total weight of acrylamide monomer and cross-linker), 3% C (amount of the crosslinker) stacking gel above a continuous 10 to 15% gradient gel (2% C) and a buffer system of 0.112 M acetate, 0.112 M Tris, pH 6.4.
  • the SDS buffer strips contain 0.20M tricine, 0.20 M Tris, 0.55% SDS at pH 7.5 in a 2% agarose gel.
  • Equal volumes of sample and 2x sample buffer (5% w/v sodium dodecyl sulfate, 0.02% bromophenol blue, with or without 10% v/v 2-mercaptoethanol) were heated for 5 minutes in a boiling water bath and 1 ul of each applied to a sample lane.
  • Low molecular weight markers (Bio-Rad cat. no. 161-0304) were included on each gel. The gels were run for 60-65 Vhr with the limiting conditions of 250V, 10 mA and 3.0 W.
  • the gels were stained in the PhastSystem development unit first with Coomassie Blue, then with silver nitrate using the PhastGel silver stain kit (Pharmacia). Gels were stained for 8 min. in. 0.1% w/v Phast Gel Blue R in 30% v/v methanol, 10% v/v acetic acid for 5 min., then switched to fresh destain for 8 min., then switched again to fresh destain for 10 min. Gels were preserved in 5% v/v glycerol, 10 v/v acetic acid for 5 min. Each gel was then washed for 2 min.
  • the product specification is that the sample must conform to standard, in this case, the untreated control.
  • Isoelectric focusing was performed using Pharmacia PhastGel IEF 3-9 precast gels of range 3 to 9 pH units.
  • the gels are approximately 0.5mm thick homogeneous polyacrylamide gels (5% T, 3%C) containing Pharmalyte carrier ampholytes.
  • the gels were prefocused for 75 Vhr using the limiting conditions of 2000V, 2.5 mA and 3.5 W.
  • the gels were then run for 410 Vhr using the same limiting conditions as during prefocusing.
  • IEF gels were stained in the PhastSystem development module as described under SDS-PAGE.
  • the product specification is that the sample must conform to standard, in this case the untreated control.
  • This affinity column was prepared by the attachment of myosin purified from dog heart tissue to cyanogen-bromide activated Sepharose®C1-4B (Pharmacia). The column was eluted with ten 1 ml aliquots of PBS-1% BSA, followed by ten 1 mL aliquots of 0.1 M glycine pH 2.5, 0.01% thimerosal. The collected fractions were counted in a gamma counter (LKB model 1272) set for In-111, the percentage eluting with the glycine buffer representing active radiolabeled antibody. This percentage was divided by the fraction of In-111 protein bound (from ITLC-SG chromatography) to correct for unbound In-111. The product specification is greater than 85% immunoreactivity.
  • the antimyosin Fab-DTPA remaining in each vial after particle counting (0.5 ml) was filtered by centrifugation through an 0.2 micron polyvinylidene difluoride filter. This method was chosen in order to allow evaluation of both the filtrate and particulates.
  • formulation development was also performed using a second murine monoclonal antibody Fab fragment, antifibrin, conjugated to the metal chelator DTPA.
  • the formulations which showed the best results were based on sodium phosphate, sodium chloride and maltose, and had a pH in the range of 6.0 to 7.2.
  • Antifibrin Fab-DTPA (Centocor, Inc.) was formulated at a protein concentration of 0.5 mg/ml into buffer solutions containing 10 mM sodium phosphate, 145 mM sodium chloride and 10% w/v maltose at pH values of 7.20, 7.00, 6.50, 6.25, 6.00 by equilibrium dialysis as described in Example 1, and vialed at 0.8 ml per vial. These vials were shipped at ambient temperature and visually inspected. The results, shown in Table 10, indicate that the formation of particulates in this formulation appeared to be pH dependent.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Genetics & Genomics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Inorganic Chemistry (AREA)
  • Biophysics (AREA)
  • Mycology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Dermatology (AREA)
  • Biomedical Technology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicinal Preparation (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Claims (13)

  1. Wäßrige Pufferlösung für monoklonale Antikörper oder monoklonale Antikörper-Fragmente, wobei die Pufferlösung aufweist:
    (a) einen Phosphatpuffer;
    (b) Natriumchlorid;
    (c) Maltose; und
    (d) monoklonale Antikörper oder monoklonale Antikörper-Fragmente.
  2. Wäßrige Pufferlösung nach Anspruch 1, wobei die Phosphatpufferlösung Natriumphosphat aufweist, das eine Konzentration zwischen 10 mM und 100 mM und einen pH-Wert zwischen 6 und 8 hat.
  3. Wäßrige Pufferlösung nach Anspruch 2, die zwischen 5 und 20 % (Gew./Vol.), beispielsweise 10 %, Maltose aufweist.
  4. Wäßrige Pufferlösung nach Anspruch 1, wobei die monoklonalen Antikörper Antimyosin-Antikörpermoleküle oder -fragmente sind.
  5. Wäßrige Pufferlösung nach Anspruch 4, wobei die monoklonalen Antikörper mit einem Chelatbildner konjugiert sind und der Chelatbildner beispielsweise DTPA ist.
  6. Wäßrige Pufferlösung nach Anspruch 1, wobei die monoklonalen Antikörper Antifibrin-Antikörpermoleküle oder -fragmente sind, die fakultativ mit einem Chelatbildner, beispielsweise DTPA, konjugiert sind.
  7. Verbesserte wäßrige Lösung nach Anspruch 1, die monoklonale Antimyosin- oder Antifibrin-Antikörper enthält und die Natriumphosphatpuffer und Natriumchlorid enthält, wobei die Verbesserung folgendes aufweist: Einbringen von zwischen 5 und 20 Gew.-% Maltose in die Lösung, wobei die Stabilität des Antikörpers in Lösung durch die Anwesenheit der Maltose erhöht wird.
  8. Wäßrige Pufferlösung für monoklonale Antikörper, die aufweist:
    (a) 10 mM bis 100 mM Natriumphosphat mit einem pH-Wert zwischen 6 und 8;
    (b) 145 mM Natriumchlorid;
    (c) 5-10 % (Gew./Vol.) Maltose; und
    (d) 0,5 bis ca. 5,2 mg/ml eines monoklonalen Antikörper-Fab-Fragments, das mit einem Metallchelator, beispielsweise DTPA, konjugiert ist.
  9. Wäßrige Pufferlösung nach Anspruch 8, wobei der Antikörper für Cardio-Myosin oder -Fibrin spezifisch ist.
  10. Wäßrige Pufferlösung nach Anspruch 8, die aufweist:
    (a) 10 mM Natriumphosphat;
    (b) 145 mM Natriumchlorid;
    (c) 10 % (Gew./Vol.) Maltose; und
    (d) 0,5 mg/ml Fab-DTPA, das für Cardio-Myosin spezifisch ist.
  11. Wäßrige Pufferlösung nach Anspruch 10, wobei das Natriumphosphat einen pH-Wert von 6,0-6,75 hat.
  12. Wäßrige Pufferlösung nach Anspruch 8, die aufweist:
    (a) 10 mM Natriumphosphat;
    (b) 145 mM Natriumchlorid;
    (c) 10 % (Gew./Vol.) Maltose; und
    (d) 0,5 mg/ml Fab-DTPA, das für Fibrin spezifisch ist.
  13. Wäßrige Pufferlösung nach Anspruch 10, 11 oder 12, wobei das Natriumphosphat einen pH-Wert zwischen 6,0 und 6,25 hat.
EP89906915A 1988-05-27 1989-05-25 Formulierung für antikörper enthaltende reagenzien Expired - Lifetime EP0417191B1 (de)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AT89906915T ATE86502T1 (de) 1988-05-27 1989-05-25 Formulierung fuer antikoerper enthaltende reagenzien.

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US19993688A 1988-05-27 1988-05-27
US199936 1998-11-25

Publications (2)

Publication Number Publication Date
EP0417191A1 EP0417191A1 (de) 1991-03-20
EP0417191B1 true EP0417191B1 (de) 1993-03-10

Family

ID=22739629

Family Applications (1)

Application Number Title Priority Date Filing Date
EP89906915A Expired - Lifetime EP0417191B1 (de) 1988-05-27 1989-05-25 Formulierung für antikörper enthaltende reagenzien

Country Status (3)

Country Link
EP (1) EP0417191B1 (de)
JP (1) JPH03504499A (de)
WO (1) WO1989011298A1 (de)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8795670B2 (en) 2002-08-16 2014-08-05 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-alpha associated disorders
US8846046B2 (en) 2002-10-24 2014-09-30 Abbvie Biotechnology Ltd. Low dose methods for treating disorders in which TNFα activity is detrimental
CN109195629A (zh) * 2016-02-24 2019-01-11 威特拉公司 流行性感冒抗体分子制剂
US10513553B2 (en) 2015-11-13 2019-12-24 Visterra, Inc. Compositions and methods for treating and preventing influenza
US10800835B2 (en) 2012-05-10 2020-10-13 Visterra, Inc. HA binding agents
US11230593B2 (en) 2019-03-25 2022-01-25 Visterra, Inc. Compositions and methods for treating and preventing influenza

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE4007079A1 (de) * 1990-03-07 1991-09-12 Behringwerke Ag Monoklonale antikoerper gegen komplexierte und nicht komplexierte komplexbildner zur entfernung von schwermetallen aus waessrigen loesungen
JPH0565233A (ja) * 1991-03-08 1993-03-19 Mitsui Toatsu Chem Inc モノクローナル抗体含有凍結乾燥製剤
JP4035167B2 (ja) 1996-02-19 2008-01-16 ヤマサ醤油株式会社 血管障害性疾患用診断薬
WO1997029785A1 (fr) * 1996-02-19 1997-08-21 Yamasa Corporation Agent d'aide au diagnostic des maladies du systeme digestif
EP0962467A4 (de) 1996-09-26 2002-10-30 Chugai Pharmaceutical Co Ltd Antikörper gegen menschliches parathormon verwandten peptide
EP0852951A1 (de) 1996-11-19 1998-07-15 Roche Diagnostics GmbH Stabile lyophilisierte pharmazeutische Zubereitungen von mono- oder polyklonalen Antikörpern
ATE361099T1 (de) 1997-05-15 2007-05-15 Chugai Pharmaceutical Co Ltd Heilmittel für kachexie
WO1999037329A1 (en) * 1998-01-22 1999-07-29 Astrazeneca Ab Pharmaceutical formulation comprising an antibody and a citrate buffer
TWI255718B (en) 1999-07-02 2006-06-01 Chugai Pharmaceutical Co Ltd Ameliorative agent for low vasopressin concentration
US20030124119A1 (en) * 1999-12-28 2003-07-03 Tadao Yamazaki Stable antibody compositions and injection preparations
EP1283057B2 (de) 2000-04-28 2012-05-30 Chugai Seiyaku Kabushiki Kaisha Zellproliferation-inhibitoren
GB0113179D0 (en) 2001-05-31 2001-07-25 Novartis Ag Organic compounds
JP4728948B2 (ja) * 2003-02-10 2011-07-20 エラン ファーマシューティカルズ,インコーポレイテッド 免疫グロブリン製剤およびその調製の方法
EP1698640B2 (de) 2003-10-01 2019-06-19 Kyowa Hakko Kirin Co., Ltd. Verfahren zur stabilisierung von antikörpern und stabilisierte antikörperzubereitung vom lösungstyp
US7785595B2 (en) 2005-04-18 2010-08-31 Yeda Research And Development Company Limited Stabilized anti-hepatitis B (HBV) antibody formulations
US8883146B2 (en) 2007-11-30 2014-11-11 Abbvie Inc. Protein formulations and methods of making same
SI3384049T1 (sl) 2015-12-03 2023-10-30 Regeneron Pharmaceuticals, Inc. Postopki povezovanja genetskih različic s kliničnim izidom pri bolnikih s starostno degeneracijo makule, zdravljenih s proti-VEGF
GB201717966D0 (en) * 2017-10-31 2017-12-13 Xenikos Bv Immunotoxins, formulations thereof and their use in medicine

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4186129A (en) * 1976-12-03 1980-01-29 Schering Aktiengesellschaft 5-(Substituted phenyl)-oxazolidinones and their sulfur analogs
EP0073371A2 (de) * 1981-08-24 1983-03-09 Miles Laboratories, Inc. Intravenös injektierbares Globulin aus Immunserum und Verfahren zu seiner Herstellung

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS597693B2 (ja) * 1978-01-07 1984-02-20 株式会社ミドリ十字 抗トロンビン製剤及びその製法
EP0124018B1 (de) * 1983-04-28 1987-11-25 Armour Pharmaceutical Company Pharmazeutische Zubereitung mit Gehalt an gereinigtem Fibrinonectin

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4186129A (en) * 1976-12-03 1980-01-29 Schering Aktiengesellschaft 5-(Substituted phenyl)-oxazolidinones and their sulfur analogs
EP0073371A2 (de) * 1981-08-24 1983-03-09 Miles Laboratories, Inc. Intravenös injektierbares Globulin aus Immunserum und Verfahren zu seiner Herstellung

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9114166B2 (en) 2002-08-16 2015-08-25 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US9302011B2 (en) 2002-08-16 2016-04-05 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-α associated disorders
US8795670B2 (en) 2002-08-16 2014-08-05 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-alpha associated disorders
US8802101B2 (en) 2002-08-16 2014-08-12 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US9220781B2 (en) 2002-08-16 2015-12-29 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US8911741B2 (en) 2002-08-16 2014-12-16 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-alpha associated disorders
US8916157B2 (en) 2002-08-16 2014-12-23 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US8916158B2 (en) 2002-08-16 2014-12-23 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US8932591B2 (en) 2002-08-16 2015-01-13 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US9272042B2 (en) 2002-08-16 2016-03-01 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US8802102B2 (en) 2002-08-16 2014-08-12 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US9327032B2 (en) 2002-08-16 2016-05-03 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US8940305B2 (en) 2002-08-16 2015-01-27 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-α associated disorders
US9272041B2 (en) 2002-08-16 2016-03-01 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US9289497B2 (en) 2002-08-16 2016-03-22 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US9295725B2 (en) 2002-08-16 2016-03-29 Abbvie Biotechnology Ltd Formulation of human antibodies for treating TNF-alpha associated disorders
US8802100B2 (en) 2002-08-16 2014-08-12 Abbvie Biotechnology Ltd. Formulation of human antibodies for treating TNF-alpha associated disorders
US8846046B2 (en) 2002-10-24 2014-09-30 Abbvie Biotechnology Ltd. Low dose methods for treating disorders in which TNFα activity is detrimental
US12024552B2 (en) 2012-05-10 2024-07-02 Visterra, Inc. Ha binding agents
US10800835B2 (en) 2012-05-10 2020-10-13 Visterra, Inc. HA binding agents
US10513553B2 (en) 2015-11-13 2019-12-24 Visterra, Inc. Compositions and methods for treating and preventing influenza
CN109195629A (zh) * 2016-02-24 2019-01-11 威特拉公司 流行性感冒抗体分子制剂
US11230593B2 (en) 2019-03-25 2022-01-25 Visterra, Inc. Compositions and methods for treating and preventing influenza

Also Published As

Publication number Publication date
EP0417191A1 (de) 1991-03-20
JPH03504499A (ja) 1991-10-03
WO1989011298A1 (en) 1989-11-30

Similar Documents

Publication Publication Date Title
EP0417191B1 (de) Formulierung für antikörper enthaltende reagenzien
US4036945A (en) Composition and method for determining the size and location of myocardial infarcts
EP1003560B1 (de) Strahlenschutzsubstanz fur radioisotopen markierte peptiden
US6024938A (en) Lyophilized imaging agent formulation comprising a chemotactic peptide
Antony et al. Isolation and characterization of a folate receptor from human placenta.
EP0417193B1 (de) Gefriergetrocknete formulierung für antikörperprodukte
Spiegelberg et al. Catabolism of human γG-immunoglobulins of different heavy chain subclasses: I. Catabolism of γG-myeloma proteins in man
Coe et al. Hamster female protein. A new Pentraxin structurally and functionally similar to C-reactive protein and amyloid P component.
US3714345A (en) Stabilized erythrocyees and methods therefore
US4636380A (en) Novel physiologic chemical method of labeling protein substances with the radionuclides of indium
CA2104772A1 (en) Method for isolating biomolecules with linear polymers
EP0028092A2 (de) Zusammensetzung, die einen radiomarkierten Antikörper enthält, Ausrüstung zu ihrer Herstellung und Ausrüstung zur Krebs- oder Tumorbestimmung, die diese Zusammensetzung enthält
CA2320156A1 (en) Reduced antigenic cells and uses therefor
Pressman et al. The nature of the combining sites of antibodies: the specific protection of the combining site by hapten during iodination
Carpenter et al. Synthetic Polypeptide Metabolism: III. Degradation and Organ Localization of Isomeric Synthetic Polypeptide Antigens
Challacombe et al. Estimation of the intravascular half-lives of normal rhesus monkey IgG, IgA and IgM.
Arnqvist et al. Hypoglycaemia caused by atypical insulin antibodies in a patient with benign monoclonal gammopathy
Bendayan et al. Influence of age on serum protein binding of propranolol
Takeda Studies of the metabolism and distribution of prothrombin in healthy men with homologous 125I-prothrombin
Tolo Penetration of human albumin through the oral mucosa of guinea-pigs immunized to this protein
DE68905336T2 (de) Formulierung fuer antikoerper enthaltende reagenzien.
Cavallo et al. Repeated exposure to bacterial lipopolysaccharide interferes with disposal of pathogenic immune complexes in mice
Thakur Immunoscintigraphic imaging of inflammatory lesions: preliminary findings and future possibilities
Spaeth et al. An Extended Clq‐Binding Assay Using Lactoperoxidase‐and Chloramine‐T‐Iodinated Clq: Immediate Distinction between Immune‐Aggregate‐Mediated and Non‐Immune‐Aggregate‐Mediated Clq Binding
Cohenford et al. Nonenzymatic glycosylation of human IgG: in vitro preparation

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19901120

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE FR GB IT LI LU NL SE

17Q First examination report despatched

Effective date: 19920410

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

ITF It: translation for a ep patent filed

Owner name: BARZANO' E ZANARDO MILANO S.P.A.

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE CH DE FR GB IT LI LU NL SE

REF Corresponds to:

Ref document number: 86502

Country of ref document: AT

Date of ref document: 19930315

Kind code of ref document: T

ET Fr: translation filed
REF Corresponds to:

Ref document number: 68905336

Country of ref document: DE

Date of ref document: 19930415

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed
PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SE

Payment date: 19940509

Year of fee payment: 6

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 19940526

Year of fee payment: 6

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: BE

Payment date: 19940527

Year of fee payment: 6

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 19940531

Year of fee payment: 6

Ref country code: LU

Payment date: 19940531

Year of fee payment: 6

Ref country code: AT

Payment date: 19940531

Year of fee payment: 6

EPTA Lu: last paid annual fee
EAL Se: european patent in force in sweden

Ref document number: 89906915.7

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 19950525

Ref country code: AT

Effective date: 19950525

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Effective date: 19950526

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Effective date: 19950531

Ref country code: CH

Effective date: 19950531

Ref country code: BE

Effective date: 19950531

BERE Be: lapsed

Owner name: CENTOCOR INC.

Effective date: 19950531

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Effective date: 19951201

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

NLV4 Nl: lapsed or anulled due to non-payment of the annual fee

Effective date: 19951201

EUG Se: european patent has lapsed

Ref document number: 89906915.7

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 19980423

Year of fee payment: 10

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 19980522

Year of fee payment: 10

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 19980723

Year of fee payment: 10

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 19990525

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 19990525

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20000131

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20000301

REG Reference to a national code

Ref country code: FR

Ref legal event code: ST

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20050525